1
|
Broer T, Tsintolas N, Hammond S, Helfer A, Lee J, Purkey K, DeLuca S, Khodabukus A, Bursac N. Human Myobundle Platform for Studying the Role of Notch Signaling in Satellite Cell Phenotype and Function. Adv Healthc Mater 2025; 14:e2404695. [PMID: 40123310 DOI: 10.1002/adhm.202404695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/27/2025] [Indexed: 03/25/2025]
Abstract
Notch signaling plays a pivotal role in regulating satellite cell (SC) behavior during skeletal muscle development, homeostasis, and repair. While well-characterized in mouse models, the impact of Notch signaling in human muscle tissues remains largely underexplored. Here, a 3D tissue-engineered model of human skeletal muscle ("myobundles") is utilized as an in vitro platform for temporal control and studies of Notch singaling. Myofiber-specific overexpression of the Notch ligand, DLL1, early in myobundle differentiation increases the abundance of 3D SCs and shifts their phenotype to a more quiescent-like state, along with decreasing muscle mass and function. In contrast, myofiber-specific DLL1 overexpression after one week of myobundle differentiation does not affect 3D SC abundance or muscle function, but increases transcriptomic markers of SC quiescence, confirming the temporal dependence of SC activation and self-renewal on Notch signaling activity. Finally, for the first time these studies show that even after a transient, myofiber-specific upregulation of Notch signaling in myobundles, 3D SCs expanded from these tissues can re-form functional "secondary" myobundles containing an amplified SC pool. Future studies in the described human myobundle platform are expected to aid the development of novel Notch-targeted therapies for muscular dystrophies and aging.
Collapse
Affiliation(s)
- Torie Broer
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Nick Tsintolas
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Stewart Hammond
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Abbigail Helfer
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Joonbum Lee
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Karly Purkey
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Sophia DeLuca
- Department of Cell Biology, Duke University, Durham, NC, 27708, USA
| | - Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
2
|
Van Keymeulen A. Mechanisms of Regulation of Cell Fate in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:167-184. [PMID: 39821026 DOI: 10.1007/978-3-031-70875-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
This chapter focuses on the mechanisms of regulation of cell fate in breast development, occurring mainly after birth, as well as in breast cancer. First, we will review how the microenvironment of the breast, as well as external cues, plays a crucial role in mammary gland cell specification and will describe how it has been shown to reprogram non-mammary cells into mammary epithelial cells. Then we will focus on the transcription factors and master regulators which have been established to be determinant for basal (BC) and luminal cell (LC) identity, and will describe the experiments of ectopic expression or loss of function of these transcription factors which demonstrated that they were crucial for cell fate. We will also discuss how master regulators are involved in the fate choice of LCs between estrogen receptor (ER)-positive cells and ER- cells, which will give rise to alveolar cells upon pregnancy and lactation. We will describe how oncogene expression induces reprogramming and change of fate of mammary epithelial cells before tumor appearance, which could be an essential step in tumorigenesis. Finally, we will describe the involvement of master regulators of mammary epithelial cells in breast cancer.
Collapse
Affiliation(s)
- Alexandra Van Keymeulen
- Laboratory of Stem Cells and Cancer (LSCC), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
3
|
Xiang Z, Wang Y, Ma X, Song S, He Y, Zhou J, Feng L, Yang S, Wu Y, Yu B, Xia G, Xu W, Zhao Y, Wang L. Targeting the NOTCH2/ADAM10/TCF7L2 Axis-Mediated Transcriptional Regulation of Wnt Pathway Suppresses Tumor Growth and Enhances Chemosensitivity in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405758. [PMID: 39601111 PMCID: PMC11744699 DOI: 10.1002/advs.202405758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/12/2024] [Indexed: 11/29/2024]
Abstract
Wnt/β-catenin/transcription factor (TCF) transcriptional activity plays an integral role in colorectal cancer (CRC) carcinogenesis. However, to date, no drugs targeting this pathway are used in clinical practice owing to the undesirable and serious side effects. In this study, it is found that the transcriptional regulation of Wnt pathway is activated and associated with liver metastasis in CRC. Through high-throughput screening of 24 inhibitors on 12 CRC and three colorectal organoids in this organoid living biobank, adavivint is found to exhibit anti-tumor activity and low toxicity in colorectal organoids, independent of the canonical Wnt/β-catenin signaling. Mechanistically, ADAM10 is screened as a target of adavivint to specifically regulate the protein expression of NOTCH2, which mediates the transcriptional regulation of the Wnt pathway. NOTCH2 not directly interact with TCF7-like 2 (TCF7L2), a key downstream transcriptional factor of canonical Wnt/β-catenin signaling, but directly activated the transcription of TCF7L2 and Wnt target genes, such as MYC, JUN and CCND1/2. Furthermore, use of adavivint or blockage of ADAM10/NOTCH2/TCF7L2 signaling enhances the chemosensitivity of CRC cells. Overall, this study provides a promising candidate for the development of small-molecule inhibitors and reveals a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhen Xiang
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Yiwei Wang
- Department of general surgeryShanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RdShanghai200233P. R. China
| | - Xiao Ma
- Fudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032P. R. China
| | - Shuzheng Song
- Department of Colorectal SurgeryDepartment of General SurgeryShanghai East HospitalTongji University School of Medicine150 Jimo RoadShanghai200120P. R. China
| | - Yuanqiao He
- Center of Laboratory Animal ScienceNanchang UniversityNo.999, Xuefu RoadNanchang330031P. R. China
| | - Jiamin Zhou
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Longhai Feng
- Department of Colorectal SurgeryThe Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital)1 Banshan East RoadHangzhou310022P. R. China
| | - Su Yang
- Department of Thoracic SurgeryRuijin HospitalShanghai Jiaotong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yibin Wu
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Bingran Yu
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Guangkai Xia
- Department of general surgeryShanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RdShanghai200233P. R. China
| | - Weiqi Xu
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Yiming Zhao
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Lu Wang
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| |
Collapse
|
4
|
Stojanovic M, Kalanj-Bognar S. Toll-like receptors as a missing link in Notch signaling cascade during neurodevelopment. Front Mol Neurosci 2024; 17:1465023. [PMID: 39664114 PMCID: PMC11631889 DOI: 10.3389/fnmol.2024.1465023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024] Open
Abstract
Neurodevelopment encompasses a complex series of molecular events occuring at defined time points distinguishable by the specific genetic readout and active protein machinery. Due to immense intricacy of intertwined molecular pathways, extracting and describing all the components of a single pathway is a demanding task. In other words, there is always a risk of leaving potential transient molecular partners unnoticed while investigating signaling cascades with core functions-and the very neglected ones could be the turning point in understanding the context and regulation of the signaling events. For example, signaling pathways of Notch and Toll-like receptors (TLRs) have been so far unrelated in the vast body of knowledge about neurodevelopment, however evidence from available literature points to their remarkable overlap in influence on identical molecular processes and reveals their potential functional links. Based on data demonstrating Notch and TLR structural engagement and functions during neurodevelopment, along with our description of novel molecular binding models, here we hypothesize that TLR proteins act as likely crucial components in the Notch signaling cascade. We advocate for the hypothesized role of TLRs in Notch signaling by: elaborating components and features of their pathways; reviewing their effects on fates of neural progenitor cells during neurodevelopment; proposing molecular and functional aspects of the hypothesis, along with venues for testing it. Finally, we discuss substantial indications of environmental influence on the proposed Notch-TLR system and its impact on neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Mario Stojanovic
- Laboratory for Neurochemistry and Molecular Neurobiology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Laboratory for Cell Biology and Signalling, Department for Molecular Biology, Institute Ruđer Bošković, Zagreb, Croatia
| | - Svjetlana Kalanj-Bognar
- Laboratory for Neurochemistry and Molecular Neurobiology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department for Chemistry and Biochemistry, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
5
|
Sudershan A, Younis M, Sudershan S, Kumar P. Migraine as an inflammatory disorder with microglial activation as a prime candidate. Neurol Res 2023; 45:200-215. [PMID: 36197286 DOI: 10.1080/01616412.2022.2129774] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2022]
Abstract
BACKGROUND The lower threshold of neuronal hyperexcitability has been correlated with migraines for decades but as technology has progressed, it has now become conceivable to learn more about the migraine disease. Apart from the "cortical spreading depression" and "activation of the trigeminovascular system", inflammation has been increasingly recognized as a possible pathogenic process that may have the possibility to regulate the disease severity. Microglial cells, the prime candidate of the innate immune cells of central nervous tissue, has been associated with numerous diseases; including cancer, neurodegenerative disorders, and inflammatory disorders. AIM In this review, we have attempted to link the dot of various microglial activation signaling pathways to enlighten the correlation between microglial involvement and the progression of migraine conditions. METHOD A structured survey of research articles and review of the literature was done in the electronic databases of Google Scholar, PubMed, Springer, and Elsevier until 31 December 2021. RESULT & CONCLUSION Of 1136 articles found initially and screening of 1047 records, 47 studies were included for the final review. This review concluded that inflammation and microglial overexpression as the prime candidate, plays an important role in the modulation of migraine and are responsible for the progression toward chronification. Therefore, this increases the possibility of preventing migraine development and chronification by blocking microglia overexpression.
Collapse
Affiliation(s)
- Amrit Sudershan
- Institute of Human Genetics, University of Jammu, Jammu and Kashmir 180006, India
| | - Mohd Younis
- Department of Human Genetics and Molecular Biology, Bharathair University, Coimbatore, 641046, India
| | - Srishty Sudershan
- Department of Zoology, University of Jammu, Jammu and Kashmir, 180006, India
| | - Parvinder Kumar
- Institute of Human Genetics, University of Jammu, Jammu and Kashmir 180006, India.,Department of Zoology, University of Jammu, Jammu and Kashmir, 180006, India
| |
Collapse
|
6
|
Therapeutic Targeting Notch2 Protects Bone Micro-Vasculatures from Methotrexate Chemotherapy-Induced Adverse Effects in Rats. Cells 2022; 11:cells11152382. [PMID: 35954226 PMCID: PMC9367713 DOI: 10.3390/cells11152382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/22/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023] Open
Abstract
Intensive cancer chemotherapy is well known to cause bone vasculature disfunction and damage, but the mechanism is poorly understood and there is a lack of treatment. Using a rat model of methotrexate (MTX) chemotherapy (five once-daily dosses at 0.75 mg/kg), this study investigated the roles of the Notch2 signalling pathway in MTX chemotherapy-induced bone micro-vasculature impairment. Gene expression, histological and micro-computed tomography (micro-CT) analyses revealed that MTX-induced micro-vasculature dilation and regression is associated with the induction of Notch2 activity in endothelial cells and increased production of inflammatory cytokine tumour necrosis factor alpha (TNFα) from osteoblasts (bone forming cells) and bone marrow cells. Blockade of Notch2 by a neutralising antibody ameliorated MTX adverse effects on bone micro-vasculature, both directly by supressing Notch2 signalling in endothelial cells and indirectly via reducing TNFα production. Furthermore, in vitro studies using rat bone marrow-derived endothelial cell revealed that MTX treatment induces Notch2/Hey1 pathway and negatively affects their ability in migration and tube formation, and Notch2 blockade can partially protect endothelial cell functions from MTX damage.
Collapse
|
7
|
Vázquez-Ulloa E, Lin KL, Lizano M, Sahlgren C. Reversible and bidirectional signaling of notch ligands. Crit Rev Biochem Mol Biol 2022; 57:377-398. [PMID: 36048510 DOI: 10.1080/10409238.2022.2113029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The Notch signaling pathway is a direct cell-cell communication system involved in a wide variety of biological processes, and its disruption is observed in several pathologies. The pathway is comprised of a ligand-expressing (sender) cell and a receptor-expressing (receiver) cell. The canonical ligands are members of the Delta/Serrate/Lag-1 (DSL) family of proteins. Their binding to a Notch receptor in a neighboring cell induces a conformational change in the receptor, which will undergo regulated intramembrane proteolysis (RIP), liberating the Notch intracellular domain (NICD). The NICD is translocated to the nucleus and promotes gene transcription. It has been demonstrated that the ligands can also undergo RIP and nuclear translocation, suggesting a function for the ligands in the sender cell and possible bidirectionality of the Notch pathway. Although the complete mechanism of ligand processing is not entirely understood, and its dependence on Notch receptors has not been ruled out. Also, ligands have autonomous functions beyond Notch activation. Here we review the concepts of reverse and bidirectional signalization of DSL proteins and discuss the characteristics that make them more than just ligands of the Notch pathway.
Collapse
Affiliation(s)
- Elenaé Vázquez-Ulloa
- Faculty of Science and Engineering/Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| | - Kai-Lan Lin
- Faculty of Science and Engineering/Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland
| | - Marcela Lizano
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Departamento de Medicina Genomica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Cecilia Sahlgren
- Faculty of Science and Engineering/Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
8
|
Excoffon KJDA, Avila CL, Alghamri MS, Kolawole AO. The magic of MAGI-1: A scaffolding protein with multi signalosomes and functional plasticity. Biol Cell 2022; 114:185-198. [PMID: 35389514 DOI: 10.1111/boc.202200014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 11/29/2022]
Abstract
MAGI-1 is a critical cellular scaffolding protein with over 110 different cellular and microbial protein interactors. Since the discovery of MAGI-1 in 1997, MAGI-1 has been implicated in diverse cellular functions such as polarity, cell-cell communication, neurological processes, kidney function, and a host of diseases including cancer and microbial infection. Additionally, MAGI-1 has undergone nomenclature changes in response to the discovery of an additional PDZ domain, leading to lack of continuity in the literature. We address the nomenclature of MAGI-1 as well as summarize many of the critical functions of the known interactions. Given the importance of many of the interactors, such as human papillomavirus E6, the Coxsackievirus and adenovirus receptor (CAR), and PTEN, the enhancement or disruption of MAGI-based interactions has the potential to affect cellular functions that can potentially be harnessed as a therapeutic strategy for a variety of diseases.
Collapse
Affiliation(s)
| | - Christina L Avila
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Mahmoud S Alghamri
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Abimbola O Kolawole
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| |
Collapse
|
9
|
Kadian LK, Arora M, Prasad CP, Pramanik R, Chauhan SS. Signaling pathways and their potential therapeutic utility in esophageal squamous cell carcinoma. Clin Transl Oncol 2022; 24:1014-1032. [PMID: 34990001 DOI: 10.1007/s12094-021-02763-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022]
Abstract
Esophageal cancer is a complex gastrointestinal malignancy with an extremely poor outcome. Approximately 80% of cases of this malignancy in Asian countries including India are of squamous cell origin, termed Esophageal Squamous Cell Carcinoma (ESCC).The five-year survival rate in ESCC patients is less than 20%. Neo-adjuvant chemo-radiotherapy (NACRT) followed by surgical resection remains the major therapeutic strategy for patients with operable ESCC. However, resistance to NACRT and local recurrence after initial treatment are the leading cause of dismal outcomes in these patients. Therefore, an alternative strategy to promote response to the therapy and reduce the post-operative disease recurrence is highly needed. At the molecular level, wide variations have been observed in tumor characteristics among different populations, nevertheless, several common molecular features have been identified which orchestrate disease progression and clinical outcome in the malignancy. Therefore, determination of candidate molecular pathways for targeted therapy remains the mainstream idea of focus in ESCC research. In this review, we have discussed the key signaling pathways associated with ESCC, i.e., Notch, Wnt, and Nrf2 pathways, and their crosstalk during disease progression. We further discuss the recent developments of novel agents to target these pathways in the context of targeted cancer therapy. In-depth research of the signaling pathways, gene signatures, and a combinatorial approach may help in discovering targeted therapy for ESCC.
Collapse
Affiliation(s)
- L K Kadian
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - M Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - C P Prasad
- Department of Medical Oncology (Lab), Dr. B. R. Ambedkar-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - R Pramanik
- Department of Medical Oncology, Dr. B. R. Ambedkar-IRCH, All India Institute of Medical Sciences, New Delhi, India
| | - S S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
10
|
Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther 2022; 7:131. [PMID: 35459215 PMCID: PMC9033871 DOI: 10.1038/s41392-022-00955-7] [Citation(s) in RCA: 500] [Impact Index Per Article: 166.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease driven by traditional and nontraditional risk factors. Genome-wide association combined with clonal lineage tracing and clinical trials have demonstrated that innate and adaptive immune responses can promote or quell atherosclerosis. Several signaling pathways, that are associated with the inflammatory response, have been implicated within atherosclerosis such as NLRP3 inflammasome, toll-like receptors, proprotein convertase subtilisin/kexin type 9, Notch and Wnt signaling pathways, which are of importance for atherosclerosis development and regression. Targeting inflammatory pathways, especially the NLRP3 inflammasome pathway and its regulated inflammatory cytokine interleukin-1β, could represent an attractive new route for the treatment of atherosclerotic diseases. Herein, we summarize the knowledge on cellular participants and key inflammatory signaling pathways in atherosclerosis, and discuss the preclinical studies targeting these key pathways for atherosclerosis, the clinical trials that are going to target some of these processes, and the effects of quelling inflammation and atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
11
|
Francis CR, Kushner EJ. Trafficking in blood vessel development. Angiogenesis 2022; 25:291-305. [PMID: 35449244 PMCID: PMC9249721 DOI: 10.1007/s10456-022-09838-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/03/2022] [Indexed: 02/17/2023]
Abstract
Blood vessels demonstrate a multitude of complex signaling programs that work in concert to produce functional vasculature networks during development. A known, but less widely studied, area of endothelial cell regulation is vesicular trafficking, also termed sorting. After moving through the Golgi apparatus, proteins are shuttled to organelles, plugged into membranes, recycled, or degraded depending on the internal and extrinsic cues. A snapshot of these protein-sorting systems can be viewed as a trafficking signature that is not only unique to endothelial tissue, but critically important for blood vessel form and function. In this review, we will cover how vesicular trafficking impacts various aspects of angiogenesis, such as sprouting, lumen formation, vessel stabilization, and secretion, emphasizing the role of Rab GTPase family members and their various effectors.
Collapse
Affiliation(s)
- Caitlin R Francis
- Department of Biological Sciences, University of Denver, Denver, CO, 80210, USA
| | - Erich J Kushner
- Department of Biological Sciences, University of Denver, Denver, CO, 80210, USA.
| |
Collapse
|
12
|
The role of A Disintegrin and Metalloproteinase (ADAM)-10 in T helper cell biology. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119192. [PMID: 34982961 DOI: 10.1016/j.bbamcr.2021.119192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
A Disintegrin and Metalloproteinases (ADAM)-10 is a member of a family of membrane-anchored proteinases that regulate a broad range of cellular functions with central roles within the immune system. This has spurred the interest to modulate ADAM activity therapeutically in immunological diseases. CD4 T helper (Th) cells are the key regulators of adaptive immune responses. Their development and function is strongly dependent on Notch, a key ADAM-10 substrate. However, Th cells rely on a variety of additional ADAM-10 substrates regulating their functional activity at multiple levels. The complexity of both, the ADAM substrate expression as well as the functional consequences of ADAM-mediated cleavage of the various substrates complicates the analysis of cell type specific effects. Here we provide an overview on the major ADAM-10 substrates relevant for CD4 T cell biology and discuss the potential effects of ADAM-mediated cleavage exemplified for a selection of important substrates.
Collapse
|
13
|
Rahn S, Becker-Pauly C. Meprin and ADAM proteases as triggers of systemic inflammation in sepsis. FEBS Lett 2022; 596:534-556. [PMID: 34762736 DOI: 10.1002/1873-3468.14225] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022]
Abstract
Systemic inflammatory disorders (SIDs) comprise a broad range of diseases characterized by dysregulated excessive innate immune responses. Severe forms of SIDs can lead to organ failure and death, and their increasing incidence represents a major issue for the healthcare system. Protease-mediated ectodomain shedding of cytokines and their receptors represents a central mechanism in the regulation of inflammatory responses. The metalloprotease A disintegrin and metalloproteinase (ADAM) 17 is the best-characterized ectodomain sheddase capable of releasing TNF-α and soluble IL-6 receptor, which are decisive factors of systemic inflammation. Recently, meprin metalloproteases were also identified as IL-6 receptor sheddases and activators of the pro-inflammatory cytokines IL-1β and IL-18. In different mouse models of SID, particularly those mimicking a sepsis-like phenotype, ADAM17 and meprins have been found to promote disease progression. In this review, we summarize the role of ADAM10, ADAM17, and meprins in the onset and progression of sepsis and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Sascha Rahn
- Biochemical Institute, Christian-Albrechts-University Kiel, Germany
| | | |
Collapse
|
14
|
Cheng XS, Shi FX, Zhao KP, Lin W, Li XY, Zhang J, Bu YY, Zhu R, Li XH, Duan DX, Ji XY, Wei JS, Wang JZ, Du J, Zhou XW. Nmnat2 attenuates amyloidogenesis and up-regulates ADAM10 in AMPK activity-dependent manner. Aging (Albany NY) 2021; 13:23620-23636. [PMID: 34644262 PMCID: PMC8580354 DOI: 10.18632/aging.203634] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/03/2021] [Indexed: 12/16/2022]
Abstract
Amyloid-β (Aβ) accumulating is considered as a causative factor for formation of senile plaque in Alzheimer’s disease (AD), but its mechanism is still elusive. The Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2), a key redox cofactor for energy metabolism, is reduced in AD. Accumulative evidence has shown that the decrease of α-secretase activity, a disintegrin and metalloprotease domain 10 (ADAM10), is responsible for the increase of Aβ productions in AD patient’s brain. Here, we observe that the activity of α-secretase ADAM10 and levels of Nmnat2 are significantly decreased, meanwhile there is a simultaneous elevation of Aβ in Tg2576 mice. Over-expression of Nmnat2 increases the mRNA expression of α-secretase ADAM10 and its activity and inhibits Aβ production in N2a/APPswe cells, which can be abolished by Compound C, an AMPK antagonist, suggesting that AMPK is involved in over-expression of Nmnat2 against Aβ production. The further assays demonstrate that Nmnat2 activates AMPK by up-regulating the ratio of NAD+/NADH, moreover AMPK agonist AICAR can also increase ADAM10 activity and reduces Aβ1-40/1-42. Taken together, Nmnat2 suppresses Aβ production and up-regulates ADAM10 in AMPK activity-dependent manner, suggesting that Nmnat2 may serve as a new potential target in arresting AD.
Collapse
Affiliation(s)
- Xiang-Shu Cheng
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China.,Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Brain Research Laboratory, Henan University, Kaifeng 475004, Henan, China
| | - Fang-Xiao Shi
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Kun-Peng Zhao
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Department of Psychiatry, Henan Key Lab of Biological Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, Henan, China
| | - Wang Lin
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xiao-Ying Li
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China
| | - Jun Zhang
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China
| | - Yao-Yao Bu
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China
| | - Rui Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xiao-Hong Li
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Dong-Xiao Duan
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Department of Physiology, Basic Medical College, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xin-Ying Ji
- Department of Microbiology, Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan, China
| | - Jian-She Wei
- Brain Research Laboratory, Henan University, Kaifeng 475004, Henan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jin Du
- Department of Neurology, Translational Medicine Center, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China.,Department of Respiratory, Huaihe Hospital Affiliated to Henan University, Kaifeng 475000, Henan, China.,Brain Research Laboratory, Henan University, Kaifeng 475004, Henan, China
| | - Xin-Wen Zhou
- Department of Pathophysiology, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Institute for Brain Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| |
Collapse
|
15
|
Okubo Y, Ohtake F, Igarashi K, Yasuhiko Y, Hirabayashi Y, Saga Y, Kanno J. Cleaved Delta like 1 intracellular domain regulates neural development via Notch signal-dependent and -independent pathways. Development 2021; 148:272156. [PMID: 34519339 PMCID: PMC8513606 DOI: 10.1242/dev.193664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/06/2021] [Indexed: 11/20/2022]
Abstract
Notch-Delta signaling regulates many developmental processes, including tissue homeostasis and maintenance of stem cells. Upon interaction of juxtaposed cells via Notch and Delta proteins, intracellular domains of both transmembrane proteins are cleaved and translocate to the nucleus. Notch intracellular domain activates target gene expression; however, the role of the Delta intracellular domain remains elusive. Here, we show the biological function of Delta like 1 intracellular domain (D1ICD) by modulating its production. We find that the sustained production of D1ICD abrogates cell proliferation but enhances neurogenesis in the developing dorsal root ganglia (DRG), whereas inhibition of D1ICD production promotes cell proliferation and gliogenesis. D1ICD acts as an integral component of lateral inhibition mechanism by inhibiting Notch activity. In addition, D1ICD promotes neurogenesis in a Notch signaling-independent manner. We show that D1ICD binds to Erk1/2 in neural crest stem cells and inhibits the phosphorylation of Erk1/2. In summary, our results indicate that D1ICD regulates DRG development by modulating not only Notch signaling but also the MAP kinase pathway.
Collapse
Affiliation(s)
- Yusuke Okubo
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Fumiaki Ohtake
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan.,Institute for Advanced Life Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Katsuhide Igarashi
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan.,Life Science Tokyo Advanced Research center (L-StaR), Hoshi University School of Pharmacy and Pharmaceutical Science, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yukuto Yasuhiko
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Yoko Hirabayashi
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Yumiko Saga
- Division of Mammalian Development, National Institute of Genetics, Yata 1111, Mishima 411-8540, Japan.,Department of Biological Science, Graduate School of Science, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jun Kanno
- Division of Cellular and Molecular Toxicology, Center for Biological Safety & Research, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| |
Collapse
|
16
|
Alghamri MS, Sharma P, Williamson TL, Readler JM, Yan R, Rider SD, Hostetler HA, Cool DR, Kolawole AO, Excoffon KJDA. MAGI-1 PDZ2 Domain Blockade Averts Adenovirus Infection via Enhanced Proteolysis of the Apical Coxsackievirus and Adenovirus Receptor. J Virol 2021; 95:e0004621. [PMID: 33762416 PMCID: PMC8437357 DOI: 10.1128/jvi.00046-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/18/2021] [Indexed: 12/14/2022] Open
Abstract
Adenoviruses (AdVs) are etiological agents of gastrointestinal, heart, eye, and respiratory tract infections that can be lethal for immunosuppressed people. Many AdVs use the coxsackievirus and adenovirus receptor (CAR) as a primary receptor. The CAR isoform resulting from alternative splicing that includes the eighth exon, CAREx8, localizes to the apical surface of polarized epithelial cells and is responsible for the initiation of AdV infection. We have shown that the membrane level of CAREx8 is tightly regulated by two MAGI-1 PDZ domains, PDZ2 and PDZ4, resulting in increased or decreased AdV transduction, respectively. We hypothesized that targeting the interactions between the MAGI-1 PDZ2 domain and CAREx8 would decrease the apical CAREx8 expression level and prevent AdV infection. Decoy peptides that target MAGI-1 PDZ2 were synthesized (TAT-E6 and TAT-NET1). PDZ2 binding peptides decreased CAREx8 expression and reduced AdV transduction. CAREx8 degradation was triggered by the activation of the regulated intramembrane proteolysis (RIP) pathway through a disintegrin and metalloproteinase (ADAM17) and γ-secretase. Further analysis revealed that ADAM17 interacts directly with the MAGI-1 PDZ3 domain, and blocking the PDZ2 domain enhanced the accessibility of ADAM17 to the substrate (CAREx8). Finally, we validated the efficacy of TAT-PDZ2 peptides in protecting the epithelia from AdV transduction in vivo using a novel transgenic animal model. Our data suggest that TAT-PDZ2 binding peptides are novel anti-AdV molecules that act by enhanced RIP of CAREx8 and decreased AdV entry. This strategy has additional translational potential for targeting other viral receptors that have PDZ binding domains, such as the angiotensin-converting enzyme 2 receptor. IMPORTANCE Adenovirus is a common threat in immunosuppressed populations and military recruits. There are no currently approved treatments/prophylactic agents that protect from most AdV infections. Here, we developed peptide-based small molecules that can suppress AdV infection of polarized epithelia by targeting the AdV receptor, coxsackievirus and adenovirus receptor (CAREx8). The newly discovered peptides target a specific PDZ domain of the CAREx8-interacting protein MAGI-1 and decrease AdV transduction in multiple polarized epithelial models. Peptide-induced CAREx8 degradation is triggered by extracellular domain (ECD) shedding through ADAM17 followed by γ-secretase-mediated nuclear translocation of the C-terminal domain. The enhanced shedding of the CAREx8 ECD further protected the epithelium from AdV infection. Taken together, these novel molecules protect the epithelium from AdV infection. This approach may be applicable to the development of novel antiviral molecules against other viruses that use a receptor with a PDZ binding domain.
Collapse
Affiliation(s)
- Mahmoud S. Alghamri
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Priyanka Sharma
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | | | - James M. Readler
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Ran Yan
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - S. Dean Rider
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, Ohio, USA
| | - Heather A. Hostetler
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, Ohio, USA
| | - David R. Cool
- Department of Pharmacology and Toxicology, Wright State University, Dayton, Ohio, USA
| | | | | |
Collapse
|
17
|
Ng HL, Quail E, Cruickshank MN, Ulgiati D. To Be, or Notch to Be: Mediating Cell Fate from Embryogenesis to Lymphopoiesis. Biomolecules 2021; 11:biom11060849. [PMID: 34200313 PMCID: PMC8227657 DOI: 10.3390/biom11060849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/29/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Notch signaling forms an evolutionarily conserved juxtacrine pathway crucial for cellular development. Initially identified in Drosophila wing morphogenesis, Notch signaling has since been demonstrated to play pivotal roles in governing mammalian cellular development in a large variety of cell types. Indeed, abolishing Notch constituents in mouse models result in embryonic lethality, demonstrating that Notch signaling is critical for development and differentiation. In this review, we focus on the crucial role of Notch signaling in governing embryogenesis and differentiation of multiple progenitor cell types. Using hematopoiesis as a diverse cellular model, we highlight the role of Notch in regulating the cell fate of common lymphoid progenitors. Additionally, the influence of Notch through microenvironment interplay with lymphoid cells and how dysregulation influences disease processes is explored. Furthermore, bi-directional and lateral Notch signaling between ligand expressing source cells and target cells are investigated, indicating potentially novel therapeutic options for treatment of Notch-mediated diseases. Finally, we discuss the role of cis-inhibition in regulating Notch signaling in mammalian development.
Collapse
Affiliation(s)
- Han Leng Ng
- Centre for Haematology, Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK;
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia; (E.Q.); (M.N.C.)
| | - Elizabeth Quail
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia; (E.Q.); (M.N.C.)
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Mark N. Cruickshank
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia; (E.Q.); (M.N.C.)
| | - Daniela Ulgiati
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia; (E.Q.); (M.N.C.)
- Correspondence: ; Tel.: +61-8-6457-1076
| |
Collapse
|
18
|
Delta-like Canonical Notch Ligand 1 in Patients Following Liver Transplantation-A Secondary Analysis of a Prospective Cohort Study. Diagnostics (Basel) 2020; 10:diagnostics10110894. [PMID: 33142943 PMCID: PMC7693674 DOI: 10.3390/diagnostics10110894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/26/2020] [Accepted: 10/29/2020] [Indexed: 12/29/2022] Open
Abstract
Opportunistic bacterial infections are dreaded risks in patients following liver transplantation (LTX), even though patients receive an antibiotic prophylaxis. The timely recognition of such an infection may be delayed, as culture-based diagnostic methods are linked with a relevant gap in performance. We measured plasma concentrations of Delta-like canonical Notch ligand 1 (DLL1) in 93 adult patients at seven consecutive time points after liver transplantation and correlated the results to the occurrence of culture-proven bacterial infection or a complicated clinical course (composite endpoint of two or more complications: graft rejection or failure, acute kidney failure, acute lung injury, or 90-day mortality). Patients exhibited elevated plasma concentrations after liver transplantation over the whole 28 d observation time. Patients with bacterial infection showed increased DLL1 levels compared to patients without infection. Persistent elevated levels of DLL1 on day 7 and afterward following LTX were able to indicate patients at risk for a complicated course. Plasma levels of DLL1 following LTX may be useful to support an earlier detection of bacterial infections in combination with C-reactive protein (CRP) and procalcitonin (PCT), or they may lead to risk stratification of patients as a single marker for post-operative complications. (Clinical Trial Notation. German Clinical Trials Register: DRKS00005480).
Collapse
|
19
|
Güner G, Lichtenthaler SF. The substrate repertoire of γ-secretase/presenilin. Semin Cell Dev Biol 2020; 105:27-42. [PMID: 32616437 DOI: 10.1016/j.semcdb.2020.05.019] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 05/17/2020] [Accepted: 05/25/2020] [Indexed: 12/09/2022]
Abstract
The intramembrane protease γ-secretase is a hetero-tetrameric protein complex with presenilin as the catalytic subunit and cleaves its membrane protein substrates within their single transmembrane domains. γ-Secretase is well known for its role in Notch signalling and in Alzheimer's disease, where it catalyzes the formation of the pathogenic amyloid β (Aβ) peptide. However, in the 21 years since its discovery many more substrates and substrate candidates of γ-secretase were identified. Although the physiological relevance of the cleavage of many substrates remains to be studied in more detail, the substrates demonstrate a broad role for γ-secretase in embryonic development, adult tissue homeostasis, signal transduction and protein degradation. Consequently, chronic γ-secretase inhibition may cause significant side effects due to inhibition of cleavage of multiple substrates. This review provides a list of 149 γ-secretase substrates identified to date and highlights by which expeirmental approach substrate cleavage was validated. Additionally, the review lists the cleavage sites where they are known and discusses the functional implications of γ-secretase cleavage with a focus on substrates identified in the recent past, such as CHL1, TREM2 and TNFR1. A comparative analysis demonstrates that γ-secretase substrates mostly have a long extracellular domain and require ectodomain shedding before γ-secretase cleavage, but that γ-secretase is also able to cleave naturally short substrates, such as the B cell maturation antigen. Taken together, the list of substrates provides a resource that may help in the future development of drugs inhibiting or modulating γ-secretase activity in a substrate-specific manner.
Collapse
Affiliation(s)
- Gökhan Güner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
20
|
Xiu MX, Liu YM, Kuang BH. The Role of DLLs in Cancer: A Novel Therapeutic Target. Onco Targets Ther 2020; 13:3881-3901. [PMID: 32440154 PMCID: PMC7213894 DOI: 10.2147/ott.s244860] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/06/2020] [Indexed: 12/18/2022] Open
Abstract
Delta-like ligands (DLLs) control Notch signaling. DLL1, DLL3 and DLL4 are frequently deregulated in cancer and influence tumor growth, the tumor vasculature and tumor immunity, which play different roles in cancer progression. DLLs have attracted intense research interest as anti-cancer therapeutics. In this review, we discuss the role of DLLs in cancer and summarize the emerging DLL-relevant targeting methods to aid future studies.
Collapse
Affiliation(s)
- Meng-Xi Xiu
- Medical School of Nanchang University, Nanchang, People's Republic of China
| | - Yuan-Meng Liu
- Medical School of Nanchang University, Nanchang, People's Republic of China
| | - Bo-Hai Kuang
- Medical School of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
21
|
Enhancing α-secretase Processing for Alzheimer's Disease-A View on SFRP1. Brain Sci 2020; 10:brainsci10020122. [PMID: 32098349 PMCID: PMC7071437 DOI: 10.3390/brainsci10020122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/24/2022] Open
Abstract
Amyloid β (Aβ) peptides generated via sequential β- and γ-secretase processing of the amyloid precursor protein (APP) are major etiopathological agents of Alzheimer's disease (AD). However, an initial APP cleavage by an α-secretase, such as the a disintegrin and metalloproteinase domain-containing protein ADAM10, precludes β-secretase cleavage and leads to APP processing that does not produce Aβ. The latter appears to underlie the disease symptom-attenuating effects of a multitude of experimental therapeutics in AD animal models. Recent work has indicated that an endogenous inhibitor of ADAM10, secreted-frizzled-related protein 1 (SFRP1), is elevated in human AD brains and associated with amyloid plaques in mouse AD models. Importantly, genetic or functional attenuation of SFRP1 lowered Aβ accumulation and improved AD-related histopathological and neurological traits. Given SFRP1's well-known activity in attenuating Wnt signaling, which is also commonly impaired in AD, SFRP1 appears to be a promising therapeutic target for AD. This idea, however, needs to be addressed with care because of cancer enhancement potentials resulting from a systemic loss of SFRP1 activity, as well as an upregulation of ADAM10 activity. In this focused review, I shall discuss α-secretase-effected APP processing in AD with a focus on SFRP1, and explore the contrasting perspectives arising from the recent findings.
Collapse
|
22
|
High-Dose Radiation Increases Notch1 in Tumor Vasculature. Int J Radiat Oncol Biol Phys 2019; 106:857-866. [PMID: 31759078 PMCID: PMC8048139 DOI: 10.1016/j.ijrobp.2019.11.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/29/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE The aim of this study is to characterize the effects of high-dose radiation therapy (HDRT) on Notch signaling components of the tumor vasculature. METHODS AND MATERIALS Human umbilical vein endothelial cells monolayers were exposed to different single fraction doses of irradiation; ribonucleic acid RNA was isolated and polymerase chain reaction was performed for Notch signaling components. The vascular response to radiation therapy was examined in a xenograft model of neuroblastoma. Tumors were treated with 0 Gy, 2 Gy, and 12 Gy single fraction doses and analyzed by double immunofluorescence staining for Notch1, Notch ligands Jagged1 and Dll4, and the endothelial cell (EC) marker endomucin. To assess the role of Notch in vivo, NGP xenograft tumors expressing Fc or Notch1-1-24-decoy (a novel Notch inhibitor) were treated with 0 Gy and 12 Gy. Immunofluorescence staining for endomucin and endomucin/αSMA was performed to analyze the effect of combination treatment on tumor EC and endothelial-to-mesenchymal-transition (EndMT), respectively. RESULTS In human umbilical vein endothelial cells monolayers doses ≥8 Gy increased expression of NOTCH1, JAG1, and Notch target genes HEY1 and HEY2 as early as 6 hours after irradiation. In vivo, 12 Gy significantly increased Notch1 and Jagged1 in tumor ECs compared with 0 Gy or 2 Gy after 72 hours. Combining HDRT with Notch inhibition using the Notch1-1-24-decoy resulted in a greater loss of EC coverage of tumor vessels than HDRT alone at 6 hours and 72 hours post treatment. Notch inhibition reduced EndMT induced by HDRT, as indicated by diminished αSMA staining in ECs. CONCLUSIONS HDRT induced Notch1 expression and increased Notch1 signaling in the endothelial component of tumor vasculature, which was not observed with lower doses. This increase in Notch1 activation might protect tumor vessels from HDRT induced damage and regulate EndMT process.
Collapse
|
23
|
Camodeca C, Cuffaro D, Nuti E, Rossello A. ADAM Metalloproteinases as Potential Drug Targets. Curr Med Chem 2019; 26:2661-2689. [PMID: 29589526 DOI: 10.2174/0929867325666180326164104] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/12/2018] [Accepted: 03/12/2018] [Indexed: 01/01/2023]
Abstract
The ADAMs, together with ADAMTSs and snake venom metalloproteases (SVMPs), are members of the Adamalysin family. Differences in structural organization, functions and localization are known and their domains, catalytic or non-catalytic, show key roles in the substrate recognition and protease activity. Some ADAMs, as membrane-bound enzymes, show sheddase activity. Sheddases are key to modulation of functional proteins such as the tumor necrosis factor, growth factors, cytokines and their receptors, adhesion proteins, signaling molecules and stress molecules involved in immunity. These activities take part in the regulation of several physiological and pathological processes including inflammation, tumor growth, metastatic progression and infectious diseases. On these bases, some ADAMs are currently investigated as drug targets to develop new alternative therapies in many fields of medicine. This review will be focused on these aspects.
Collapse
Affiliation(s)
- Caterina Camodeca
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa, Italy
| | - Doretta Cuffaro
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa, Italy
| | - Elisa Nuti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa, Italy
| | - Armando Rossello
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa, Italy
| |
Collapse
|
24
|
Lee YJ, Ch'ng TH. RIP at the Synapse and the Role of Intracellular Domains in Neurons. Neuromolecular Med 2019; 22:1-24. [PMID: 31346933 DOI: 10.1007/s12017-019-08556-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022]
Abstract
Regulated intramembrane proteolysis (RIP) occurs in a cell when transmembrane proteins are cleaved by intramembrane proteases such as secretases to generate soluble protein fragments in the extracellular environment and the cytosol. In the cytosol, these soluble intracellular domains (ICDs) have local functions near the site of cleavage or in many cases, translocate to the nucleus to modulate gene expression. While the mechanism of RIP is relatively well studied, the fate and function of ICDs for most substrate proteins remain poorly characterized. In neurons, RIP occurs in various subcellular compartments including at the synapse. In this review, we summarize current research on RIP in neurons, focusing specifically on synaptic proteins where the presence and function of the ICDs have been reported. We also briefly discuss activity-driven processing of RIP substrates at the synapse and the cellular machinery that support long-distance transport of ICDs from the synapse to the nucleus. Finally, we describe future challenges in this field of research in the context of understanding the contribution of ICDs in neuronal function.
Collapse
Affiliation(s)
- Yan Jun Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, 10-01-01 M, Singapore, 308232, Singapore.,Interdisciplinary Graduate School (IGS), Nanyang Technological University, Singapore, Singapore
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, 10-01-01 M, Singapore, 308232, Singapore. .,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
25
|
Hildebrand D, Decker SO, Koch C, Schmitt FCF, Ruhrmann S, Schneck E, Sander M, Weigand MA, Brenner T, Heeg K, Uhle F. Host-Derived Delta-Like Canonical Notch Ligand 1 as a Novel Diagnostic Biomarker for Bacterial Sepsis-Results From a Combinational Secondary Analysis. Front Cell Infect Microbiol 2019; 9:267. [PMID: 31396491 PMCID: PMC6663974 DOI: 10.3389/fcimb.2019.00267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/09/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Sepsis is a life-threatening syndrome, resulting from a dysbalanced host response to infection. However, especially the early, pro-inflammatory immune response in sepsis is similar to other inflammatory conditions without infectious cause, e.g., trauma or surgery. This aspect challenges the value of current biomarkers for diagnosis, as these are often broadly induced. We earlier identified Delta-like Protein 1 (DLL1), a canonical Notch ligand, to be released from monocytes upon bacterial stimulation. Considering the importance of monocytes in the pathophysiology of sepsis, we hypothesized that this mechanism might occur also in the clinical setting and DLL1 might serve as a biomarker of life-threatening bacterial infection. Methods: We combined samples from three different studies, including subgroups of patients with sepsis (n = 80), surgical patients (n = 50), trauma patients (n = 36), as well as healthy controls (n = 50). We assessed plasma concentrations of DLL1 using ELISA. We performed Area-under-receiver-operator-curve (AUROC) analysis to evaluate the diagnostic performance of DLL1 compared to leucocytes, C-reactive protein (CRP), and procalcitonin (PCT). Results: Plasma concentrations of DLL1 were strongly elevated already at sepsis onset and maintained elevated until day 7. In contrast, neither surgical patients nor patients after severe trauma presented with elevated levels, while conventional biomarkers of inflammation (e.g., leucocytes and CRP), responded. AUROC analysis revealed a cut-off of 30 ng/ml associated with the best diagnostic performance, yielding a superior accuracy of 91% for DLL1, compared to 75, 79, and 81% for CRP, leucocytes, and PCT. Conclusion: DLL1 is a novel host-derived biomarker for the diagnosis of sepsis with a better performance compared to established ones, most likely due to its high robustness in non-infectious inflammatory responses. Clinical Trial Registration:POCSEP-Trial DRKS00008090; MIRSI DRKS00005463; SPRINT DRKS00010991.
Collapse
Affiliation(s)
- Dagmar Hildebrand
- Medical Microbiology and Hygiene, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Sebastian O Decker
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Koch
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Giessen, Germany
| | - Felix C F Schmitt
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sophie Ruhrmann
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Giessen, Germany
| | - Emmanuel Schneck
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Giessen, Germany
| | - Michael Sander
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Giessen, Germany
| | | | - Thorsten Brenner
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus Heeg
- Medical Microbiology and Hygiene, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
26
|
Xiu MX, Liu YM. The role of oncogenic Notch2 signaling in cancer: a novel therapeutic target. Am J Cancer Res 2019; 9:837-854. [PMID: 31218097 PMCID: PMC6556604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/22/2019] [Indexed: 06/09/2023] Open
Abstract
Deregulated Notch signaling is a key factor thought to facilitate the stem-like proliferation of cancer cells, thereby facilitating disease progression. Four subtypes of Notch receptor have been described to date, with each playing a distinct role in cancer development and progression, therefore warranting a careful and comprehensive examination of the targeting of each receptor subtype in the context of oncogenesis. Clinical efforts to translate the DAPT, which blocks Notch signaling, have been unsuccessful due to a combination of serious gastrointestinal side effects and a lack of complete blocking efficacy. There is therefore a clear need to identify better therapeutic strategies for targeting and manipulating Notch signaling. Notch2 is a Notch receptor that is commonly overexpressed in a range of cancers, and which is linked to a unique oncogenic mechanism. Successful efforts to block Notch2 signaling will depend upon doing so both efficiently and specifically in patients. As such, in the present review we will explore the role of Notch2 signaling in the development and progression of cancer, and we will assess agents and strategies with the potential to effectively disrupt Notch2 signaling and thereby yield novel cancer treatment regimens.
Collapse
Affiliation(s)
- Meng-Xi Xiu
- Medical School of Nanchang University Nanchang, Jiangxi, China
| | - Yuan-Meng Liu
- Medical School of Nanchang University Nanchang, Jiangxi, China
| |
Collapse
|
27
|
Abstract
Notch signaling regulates a multitude of cellular processes. During ocular lens development this pathway is required for lens progenitor growth, differentiation and maintenance of the transition zone. After ligand-receptor binding, the receptor proteins are processed, first by ADAM proteases, then by γ-secretase cleavage. This results in the release of a Notch intracellular domain (N-ICD), which is recruited into a nuclear transcription factor complex that activates Notch target genes. Previous in vitro studies showed that the Delta-like and Jagged ligand proteins can also be cleaved by the γ-secretase complex, but it remains unknown whether such processing occurs during in vivo vertebrate development. Here we show that mouse and human lens progenitor cells endogenously express multiple Jagged1 protein isoforms, including a Jagged1 intracellular domain. We also found that pharmacologic blockage of γ-secretase activity in vitro resulted in an accumulation of Jagged1 polypeptide intermediates. Finally, overexpression of an epitope-tagged Jagged1 intracellular domain displayed nuclear localization and induced the upregulation of endogenous JAG1 mRNA expression. These findings support the idea that along with its classical role as a Notch pathway ligand, Jagged1 is regulated post-translationally, to produce multiple active protein isoforms. Summary: The Notch pathway ligand protein Jagged1 undergoes multiple catalytic cleavages, regulated by Adam proteases and the gamma-secretase complex, during mammalian lens development, similar to Notch receptor proteins.
Collapse
Affiliation(s)
- Mina Azimi
- Department of Cell Biology & Human Anatomy, University of California, 1 Shields Avenue, Davis, CA 95616, USA
| | - Nadean L Brown
- Department of Cell Biology & Human Anatomy, University of California, 1 Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
28
|
Nutritional Preconditioning of Apigenin Alleviates Myocardial Ischemia/Reperfusion Injury via the Mitochondrial Pathway Mediated by Notch1/Hes1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7973098. [PMID: 31015891 PMCID: PMC6446095 DOI: 10.1155/2019/7973098] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/01/2018] [Accepted: 01/27/2019] [Indexed: 01/08/2023]
Abstract
Apigenin (Api), a natural flavone found in high amounts in several herbs, has shown potent cardioprotective effects in clinical studies, although the underlying mechanisms are not clear. We hypothesized that Api protects the myocardium from simulated ischemia/reperfusion (SI/R) injury via nutritional preconditioning (NPC). Rats fed with Api-containing food showed improvement in cardiac functions; lactate dehydrogenase (LDH) and creatine phosphokinase (CPK) activities; infarct size; apoptosis rates; malondialdehyde (MDA) levels; caspase-3, superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) activities; and ferric reducing antioxidant power (FRAP) compared to those fed standard chow following SI/R injury. In addition, Api pretreatment significantly improved the viability, decreased the LDH activity and intracellular reactive oxygen species (ROS) generation, alleviated the loss of mitochondrial membrane potential (MMP), prevented the opening of the mitochondrial permeability transition pore (mPTP), and decreased the caspase-3 activity, cytochrome c (Cyt C) release, and apoptosis induced by SI/R in primary cardiomyocytes. Mechanistically, Api upregulated Hes1 expression and was functionally neutralized by the Notch1 γ-secretase inhibitor GSI, as well as the mPTP opener atractyloside (Atr). Taken together, Api protected the myocardium against SI/R injury via the mitochondrial pathway mediated by the Notch1/Hes1 signaling pathway.
Collapse
|
29
|
ADAM10 in Alzheimer's disease: Pharmacological modulation by natural compounds and its role as a peripheral marker. Biomed Pharmacother 2019; 113:108661. [PMID: 30836275 DOI: 10.1016/j.biopha.2019.108661] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) represents a global burden in the economics of healthcare systems. Amyloid-β (Aβ) peptides are formed by amyloid-β precursor protein (AβPP) cleavage, which can be processed by two pathways. The cleavage by the α-secretase A Disintegrin And Metalloprotease 10 (ADAM10) releases the soluble portion (sAβPPα) and prevents senile plaques. This pathway remains largely unknown and ignored, mainly regarding pharmacological approaches that may act via different signaling cascades and thus stimulate non-amyloidogenic cleavage through ADAM10. This review emphasizes the effects of natural compounds on ADAM10 modulation, which eventuates in a neuroprotective mechanism. Moreover, ADAM10 as an AD biomarker is revised. New treatments and preventive interventions targeting ADAM10 regulation for AD are necessary, considering the wide variety of ADAM10 substrates.
Collapse
|
30
|
Immunoreceptor Engineering and Synthetic Cytokine Signaling for Therapeutics. Trends Immunol 2019; 40:258-272. [DOI: 10.1016/j.it.2019.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 12/25/2022]
|
31
|
Madav Y, Wairkar S, Prabhakar B. Recent therapeutic strategies targeting beta amyloid and tauopathies in Alzheimer's disease. Brain Res Bull 2019; 146:171-184. [PMID: 30634016 DOI: 10.1016/j.brainresbull.2019.01.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/13/2018] [Accepted: 01/03/2019] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) has been a global concern for years due to its severe implications that affects the quality of life of the patients. The available line of therapy for treating Alzheimer's includes acetylcholinesterase inhibitors, NMDA(N-methyl-D-aspartate) antagonists and their combination which gives only symptomatic relief rather than treating the root cause of AD. Senile plaques and neurofibrillary tangles are the characteristic features underlying Alzheimer's pathology. Several attempts have been made towards exploring the niceties of these hallmarks and targeting various aspects of amyloid and tau pathology at different stages to eliminate the ultimate cause. Approaches targeting cleavage and formation of toxic amyloid fragments by secretases, aggregation of amyloid monofilaments, and immunotherapy against amyloid deposits has been extensively studied to treat amyloid pathology. Similarly, for tau pathology, tau hyperphosphorylation, microtubule stabilization, anti-tau immunotherapy has been explored. This article focuses on AD pathology and current pharmacotherapy, precisely for amyloid and tau. Furthermore, preclinical and clinical studies along with potential leads discovered under these approaches have also been included in this article. However, despite extensive research in drug development, overcoming clinical barrier still remain a major challenge for Alzheimer's pharmacotherapy.
Collapse
Affiliation(s)
- Yamini Madav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Bala Prabhakar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
32
|
IL-4-dependent Jagged1 expression/processing is associated with survival of chronic lymphocytic leukemia cells but not with Notch activation. Cell Death Dis 2018; 9:1160. [PMID: 30478302 PMCID: PMC6255763 DOI: 10.1038/s41419-018-1185-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 12/15/2022]
Abstract
As previously reported, chronic lymphocytic leukemia (CLL) cells show constitutive Notch1/2 activation and express the Notchligand Jagged1. Despite increasing knowledge of the impact of Notch alterations on CLL biology and pathogenesis, the role of Jagged1 expressed in CLL cells remains undefined. In other cell types, it has been shown that after Notch engagement, Jagged1 not only activates Notch in signal-receiving cell, but also undergoes proteolytic activation in signal-sending cell, triggering a signaling with biological effects. We investigated whether Jagged1 expressed in CLL cells undergoes proteolytic processing and/or is able to induce Notch activation through autocrine/paracrine loops, focusing on the effect that CLL prosurvival factor IL-4 could exert on the Notch-Jagged1 system in these cells. We found that Jagged1 was constitutively processed in CLL cells and generated an intracellular fragment that translocated into the nucleus, and an extracellular fragment released into the culture supernatant. IL-4 enhanced expression of Jagged1 and its intracellular fragments, as well as Notch1/2 activation. The IL-4-induced increase in Notch1/2 activation was independent of the concomitant upregulated Jagged1 levels. Indeed, blocking Notch-Jagged1 interactions among CLL cells with Jagged1 neutralizing antibodies did not affect the expression of the Notch target Hes1. Notably, anti-Jagged1 antibodies partially prevented the IL-4-induced increase in Jagged1 processing and cell viability, suggesting that Jagged1 processing is one of the events contributing to IL-4-induced CLL cell survival. Consistent with this, Jagged1 silencing by small interfering RNA partially counteracted the capacity of IL-4 to promote CLL cell survival. Investigating the pathways whereby IL-4 promoted Notch1/2 activation in CLL cells independent of Jagged1, we found that PI3Kδ/AKT and PKCδ were involved in upregulating Notch1 and Notch2 proteins, respectively. Overall, this study provides new insights into the Notch-ligand system in CLL cells and suggests that targeting this system may be exploited as a novel/additional therapy approach for CLL.
Collapse
|
33
|
Van Sinderen M, Oyanedel J, Menkhorst E, Cuman C, Rainczuk K, Winship A, Salamonsen L, Edgell T, Dimitriadis E. Soluble Delta-like ligand 1 alters human endometrial epithelial cell adhesive capacity. Reprod Fertil Dev 2018; 29:694-702. [PMID: 26616664 DOI: 10.1071/rd15313] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/23/2015] [Indexed: 01/23/2023] Open
Abstract
The endometrium undergoes substantial morphological and functional changes to become receptive to embryo implantation and to enable establishment of a successful pregnancy. Reduced Delta-like ligand 1 (DLL1, Notch ligand) in the endometrium is associated with infertility. DLL1 can be cleaved by 'a disintegrin and metalloprotease' (ADAM) proteases to produce a soluble ligand that may act to inhibit Notch signalling. We used an enzyme-linked immunosorbent assay to quantify soluble DLL1 in uterine lavages from fertile and infertile women in the secretory phase of the menstrual cycle. We also determined the cellular location and immunostaining intensity of ADAM12 and 17 in human endometrium throughout the cycle. Functional effects of soluble DLL1 in receptivity were analysed using in vitro adhesion and proliferation assays and gene expression analysis of Notch signalling targets. Soluble DLL1 was significantly increased in uterine lavage samples of infertile women compared with fertile women in the secretory phase of the menstrual cycle. This coincided with significantly increased ADAM17 immunostaining detected in the endometrial luminal epithelium in the mid-secretory phase in infertile women. Soluble DLL1 significantly inhibited the adhesive capacity of endometrial epithelial cells via downregulation of helix-loop-helix and hairy/enhancer of split family member HES1 mRNA. Thus, soluble DLL1 may serve as a suitable target or potential biomarker for receptivity.
Collapse
Affiliation(s)
- Michelle Van Sinderen
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - Jennifer Oyanedel
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - Ellen Menkhorst
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - Carly Cuman
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - Katarzyna Rainczuk
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - Amy Winship
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - Lois Salamonsen
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - Tracey Edgell
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - Evdokia Dimitriadis
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| |
Collapse
|
34
|
Latifi Z, Fattahi A, Ranjbaran A, Nejabati HR, Imakawa K. Potential roles of metalloproteinases of endometrium-derived exosomes in embryo-maternal crosstalk during implantation. J Cell Physiol 2017; 233:4530-4545. [PMID: 29115666 DOI: 10.1002/jcp.26259] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/09/2017] [Indexed: 12/15/2022]
Abstract
During embryo implantation, crosstalk between the endometrial epithelium and the blastocyst, especially the trophoblasts, is a prerequisite for successful implantation. During this crosstalk, various molecular and functional changes occur to promote synchrony between the embryo and the endometrium as well as the uterine cavity microenvironment. In the past few years, growing evidence has shown that endometrium-derived exosomes play pivotal roles in the embryonic-maternal crosstalk during implantation, although the exact mechanism of this crosstalk has yet to be determined. The presence of metalloproteinases has been reported in endometrium-derived exosomes, implying the importance of these enzymes in exosome-based crosstalk. Thus, in this review, we describe the potential roles of the metalloproteinases of endometrium-derived exosomes in promoting embryo attachment and implantation. This study could provide a better understanding of the potential roles of exosomal metalloproteinases in embryo implantation and pave the way for developing novel exosome-based regulatory agents to support early pregnancy.
Collapse
Affiliation(s)
- Zeinab Latifi
- Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ibaraki, Japan.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Ranjbaran
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Nejabati
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kazuhiko Imakawa
- Animal Resource Science Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ibaraki, Japan
| |
Collapse
|
35
|
Gazave E, Lemaître QIB, Balavoine G. The Notch pathway in the annelid Platynereis: insights into chaetogenesis and neurogenesis processes. Open Biol 2017; 7:rsob.160242. [PMID: 28148821 PMCID: PMC5356439 DOI: 10.1098/rsob.160242] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/03/2017] [Indexed: 01/13/2023] Open
Abstract
Notch is a key signalling pathway playing multiple and varied functions during development. Notch regulates the selection of cells with a neurogenic fate and maintains a pool of yet uncommitted precursors through lateral inhibition, both in insects and in vertebrates. Here, we explore the functions of Notch in the annelid Platynereis dumerilii (Lophotrochozoa). Conserved components of the pathway are identified and a scenario for their evolution in metazoans is proposed. Unexpectedly, neither Notch nor its ligands are expressed in the neurogenic epithelia of the larva at the time when massive neurogenesis begins. Using chemical inhibitors and neural markers, we demonstrate that Notch plays no major role in the general neurogenesis of larvae. Instead, we find Notch components expressed in nascent chaetal sacs, the organs that produce the annelid bristles. Impairing Notch signalling induces defects in chaetal sac formation, abnormalities in chaetae producing cells and a change of identity of chaeta growth accessory cells. This is the first bilaterian species in which the early neurogenesis processes appear to occur without a major involvement of the Notch pathway. Instead, Notch is co-opted to pattern annelid-specific organs, likely through a lateral inhibition process. These features reinforce the view that Notch signalling has been recruited multiple times in evolution due to its remarkable ‘toolkit’ nature.
Collapse
Affiliation(s)
- Eve Gazave
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot, Sorbonne Paris Cité, 75205 Paris, France
| | - Quentin I B Lemaître
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot, Sorbonne Paris Cité, 75205 Paris, France
| | - Guillaume Balavoine
- Institut Jacques Monod, CNRS, UMR 7592, Univ Paris Diderot, Sorbonne Paris Cité, 75205 Paris, France
| |
Collapse
|
36
|
Forghany Z, Robertson F, Lundby A, Olsen JV, Baker DA. Control of endothelial cell tube formation by Notch ligand intracellular domain interactions with activator protein 1 (AP-1). J Biol Chem 2017; 293:1229-1242. [PMID: 29196606 DOI: 10.1074/jbc.m117.819045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/30/2017] [Indexed: 01/08/2023] Open
Abstract
Notch signaling is a ubiquitous signal transduction pathway found in most if not all metazoan cell types characterized to date. It is indispensable for cell differentiation as well as tissue growth, tissue remodeling, and apoptosis. Although the canonical Notch signaling pathway is well characterized, accumulating evidence points to the existence of multiple, less well-defined layers of regulation. In this study, we investigated the function of the intracellular domain (ICD) of the Notch ligand Delta-like 4 (DLL4). We provide evidence that the DLL4 ICD is required for normal DLL4 subcellular localization. We further show that it is cleaved and interacts with the JUN proto-oncogene, which forms part of the activator protein 1 (AP-1) transcription factor complex. Mechanistically, the DLL4 ICD inhibited JUN binding to DNA and thereby controlled the expression of JUN target genes, including DLL4 Our work further demonstrated that JUN strongly stimulates endothelial cell tube formation and that DLL4 constrains this process. These results raise the possibility that Notch/DLL4 signaling is bidirectional and suggest that the DLL4 ICD could represent a point of cross-talk between Notch and receptor tyrosine kinase (RTK) signaling.
Collapse
Affiliation(s)
- Zary Forghany
- From the Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands and
| | - Francesca Robertson
- From the Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands and
| | - Alicia Lundby
- Novo Nordisk Foundation Center for Protein Research and.,the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | | | - David A Baker
- From the Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands and
| |
Collapse
|
37
|
Mills EA, Goldman D. The Regulation of Notch Signaling in Retinal Development and Regeneration. CURRENT PATHOBIOLOGY REPORTS 2017; 5:323-331. [PMID: 29354328 DOI: 10.1007/s40139-017-0153-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Purpose of review Notch signaling is an important component of retinal progenitor cell maintenance and MG specification during development, and its manipulation may be critical for allowing MG to re-enter the cell cycle and regenerate neurons in adults. In mammals, MG respond to retinal injury by undergoing a gliotic response rather than a regenerative one. Understanding the complexities of Notch signaling may allow for strategies that enhance regeneration over gliosis. Recent findings Notch signaling is regulated at multiple levels, and is interdependent with various other signaling pathways in both the receptor and ligand expressing cells. The precise spatial and temporal patterning of Notch components is necessary for proper retinal development. Regenerative species undergo a dynamic regulation of Notch signaling in MG upon injury, whereas non-regenerative species fail to productively regulate Notch. Summary Notch signaling is malleable, such that the altered composition of growth and transcription factors in the developing and mature retinas result in different Notch mediated responses. Successful regeneration will require the manipulation of the retinal environment to foster a dynamic rather than static regulation of Notch signaling in concert with other reprogramming and differentiation factors.
Collapse
Affiliation(s)
- Elizabeth A Mills
- Molecular and Behavioral Neuroscience Institute, Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109
| | - Daniel Goldman
- Molecular and Behavioral Neuroscience Institute, Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
38
|
Liu T, He W, Li Y. Helicobacter pylori Infection of Gastric Epithelial Cells Affects NOTCH Pathway In Vitro. Dig Dis Sci 2016; 61:2516-21. [PMID: 27073072 DOI: 10.1007/s10620-016-4161-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 04/04/2016] [Indexed: 01/24/2023]
Abstract
BACKGROUND Helicobacter pylori infection is exceptionally prevalent, and it is an important risk factor for gastritis, gastroduodenal ulcers, and gastric cancer. However, the pathogenic mechanisms of H. pylori are not entirely clear. The aim of this study was to assess which signal pathway is initially activated by H. pylori. METHODS Using the Human Signal Transduction Pathway Finder RT(2) Profiler PCR Array, we screened for alterations in the expression of genes encoding members of ten different signal transduction pathways in GES-1 cells co-cultured with H. pylori. qPCR and Western blotting were used to verify the expression of four key genes in NOTCH pathway. RESULTS Of the 84 genes represented in the array, 22 genes demonstrated more than twofold difference (p < 0.05) in GES-1 cells grown in the presence of H. pylori 11637 compared to cells without H. pylori 11637. Ten genes were up-regulated in the co-culture group, whereas 12 appeared to be down-regulated. Further analysis using the SA Biosciences online program revealed that NOTCH pathway was the most significantly affected network. There was a significant reduction in the mRNA expression level of NOTCH1 and NOTCH2, together with a reduced level of active forms of NOTCH1 (NICD1) and NOTCH2 (NICD2). Meanwhile, the expression level of the ligand DLL4 was found to be significantly increased. CONCLUSIONS NOTCH signaling may play an important role in H. pylori-induced gastric carcinogenesis.
Collapse
Affiliation(s)
- Tao Liu
- Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu Province, China
- Key Laboratory of Digestive System Tumors, Lanzhou, Gansu Province, China
| | - Wenting He
- Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu Province, China
- Key Laboratory of Digestive System Tumors, Lanzhou, Gansu Province, China
| | - Yumin Li
- Second Hospital of Lanzhou University, Lanzhou, 730030, Gansu Province, China.
- Key Laboratory of Digestive System Tumors, Lanzhou, Gansu Province, China.
| |
Collapse
|
39
|
Tu KS, Yao YM. Epithelial-mesenchymal transition and related signaling pathways in hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2016; 24:2131-2142. [DOI: 10.11569/wcjd.v24.i14.2131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common forms of liver cancer and the third leading cause of cancer-related mortality in the world. Although numerous therapeutic strategies have been employed to treat this fatal disease, the prognosis of HCC patients remains dismal with a low 5-year survival rate of approximately 30%. Postoperative recurrence and metastasis of HCC are the leading cause of poor prognosis. Metastasis has been thought to rely on non-motile epithelial tumor cells acquiring characteristics of mesenchymal cells, which are more migratory. This change is known as the epithelial-to-mesenchymal transition (EMT). EMT has been considered one of the main reasons for the invasion and metastasis of HCC. Notably, increasing evidence indicates that several signaling pathways participate in the regulation of EMT in HCC. In the current review, we will discuss the current progress in research of EMT and its related signaling pathways in HCC.
Collapse
|
40
|
Combinatorial microenvironmental regulation of liver progenitor differentiation by Notch ligands, TGFβ, and extracellular matrix. Sci Rep 2016; 6:23490. [PMID: 27025873 PMCID: PMC4812246 DOI: 10.1038/srep23490] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/08/2016] [Indexed: 12/21/2022] Open
Abstract
The bipotential differentiation of liver progenitor cells underlies liver development and bile duct formation as well as liver regeneration and disease. TGFβ and Notch signaling are known to play important roles in the liver progenitor specification process and tissue morphogenesis. However, the complexity of these signaling pathways and their currently undefined interactions with other microenvironmental factors, including extracellular matrix (ECM), remain barriers to complete mechanistic understanding. Utilizing a series of strategies, including co-cultures and cellular microarrays, we identified distinct contributions of different Notch ligands and ECM proteins in the fate decisions of bipotential mouse embryonic liver (BMEL) progenitor cells. In particular, we demonstrated a cooperative influence of Jagged-1 and TGFβ1 on cholangiocytic differentiation. We established ECM-specific effects using cellular microarrays consisting of 32 distinct combinations of collagen I, collagen III, collagen IV, fibronectin, and laminin. In addition, we demonstrated that exogenous Jagged-1, Delta-like 1, and Delta-like 4 within the cellular microarray format was sufficient for enhancing cholangiocytic differentiation. Further, by combining Notch ligand microarrays with shRNA-based knockdown of Notch ligands, we systematically examined the effects of both cell-extrinsic and cell-intrinsic ligand. Our results highlight the importance of divergent Notch ligand function and combinatorial microenvironmental regulation in liver progenitor fate specification.
Collapse
|
41
|
Glycoprotein nonmetastatic melanoma protein B extracellular fragment shows neuroprotective effects and activates the PI3K/Akt and MEK/ERK pathways via the Na+/K+-ATPase. Sci Rep 2016; 6:23241. [PMID: 26988030 PMCID: PMC4796790 DOI: 10.1038/srep23241] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 03/02/2016] [Indexed: 12/23/2022] Open
Abstract
Glycoprotein nonmetastatic melanoma protein B (GPNMB) plays important roles in various types of cancer and amyotrophic lateral sclerosis (ALS). The details of GPNMB function and its interacting protein have not been clarified. Therefore, to identify GPNMB binding partners on the cell membrane, we used membrane protein library/BLOTCHIP-MS technology, which enables us to analyze all cell membrane proteins as binding partners of the GPNMB extracellular fragment. As a result of a comprehensive search, we identified the alpha subunits of Na(+)/K(+)-ATPase (NKA) as a possible binding partner. We confirmed the interaction between the GPNMB extracellular fragment and NKA by immunoprecipitation and immunostaining in NSC-34 cells. Indeed, endogenous GPNMB extracellular fragment bound to and colocalized with NKA alpha subunits. Furthermore, exogenous GPNMB extracellular fragment, i.e., human recombinant GPNMB, also bound to and colocalized with NKA alpha subunits. Additionally, we found that the GPNMB extracellular fragment had neuroprotective effects and activated the phosphoinositide 3-kinase (PI3K)/Akt and mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK pathways via NKA. These findings indicated that NKA may act as a novel "receptor" for the GPNMB extracellular fragment, offering additional molecular targets for the treatment of GPNMB-related diseases, including various types of cancer and ALS.
Collapse
|
42
|
Turkoz M, Townsend RR, Kopan R. The Notch Intracellular Domain Has an RBPj-Independent Role during Mouse Hair Follicular Development. J Invest Dermatol 2016; 136:1106-1115. [PMID: 26940862 DOI: 10.1016/j.jid.2016.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/15/2016] [Accepted: 02/07/2016] [Indexed: 01/02/2023]
Abstract
Ligand-dependent activation, γ-secretase-processed cleavage, and recombining binding protein Jk (RBPj)-mediated downstream transcriptional activities of Notch receptors constitute the "canonical" Notch signaling pathway, which is essential for skin organogenesis. However, in Msx2-Cre mice, keratinocyte-specific deletion of the Rbpj gene in utero produced a significantly milder phenotype than either global Notch or γ-secretase loss. Herein, we investigated the underlying mechanisms for this apparent noncanonical signal using mouse genetics. We found no evidence that ligand back-signaling contributed to skin organogenesis. The perdurance of RBPj protein did not establish an epigenetic memory of a canonical signal in the youngest epidermal stem cells, and Notch targets were not derepressed. We provide evidence that γ-secretase-dependent but RBPj-independent Notch intracellular domain activity operating in the first hair follicles is responsible for a delay in follicular destruction, which results in lower serum thymic stromal lymphopoietin levels, milder B-cell lymphoproliferative disease, and improved survival in Msx2-Cre(+/tg);Rbpj(f/f) mice. Minimal amounts of the Notch intracellular domain were sufficient for rescue, which was not mediated by transcription, suggesting that the Notch intracellular domain is acting through a novel mechanism.
Collapse
Affiliation(s)
- Mustafa Turkoz
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - R Reid Townsend
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Raphael Kopan
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, USA.
| |
Collapse
|
43
|
Lim KJ, Brandt WD, Heth JA, Muraszko KM, Fan X, Bar EE, Eberhart CG. Lateral inhibition of Notch signaling in neoplastic cells. Oncotarget 2015; 6:1666-77. [PMID: 25557173 PMCID: PMC4359323 DOI: 10.18632/oncotarget.2762] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/17/2014] [Indexed: 01/07/2023] Open
Abstract
During normal development, heterogeneous expression of Notch ligands can result in pathway suppression in the signal-sending cell, a process known as lateral inhibition. It is unclear if an analogous phenomenon occurs in malignant cells. We observed significant induction of Notch ligands in glioblastoma neurospheres and pancreatic carcinoma cells cultured in low oxygen, suggesting that this phenomenon could occur around hypoxic regions. To model lateral inhibition in these tumors, the ligand Jagged1 was overexpressed in glioblastoma and pancreatic carcinoma cells, resulting in overall induction of pathway targets. However, when ligand high and ligand low cells from a single line were co-cultured and then separated, we noted suppression of Notch pathway targets in the former and induction in the latter, suggesting that neoplastic lateral inhibition can occur. We also found that repression of Notch pathway targets in signal-sending cells may occur through the activity of a Notch ligand intracellular domain, which translocates into the nucleus. Understanding how this neoplastic lateral inhibition process functions in cancer cells may be important in targeting ligand driven Notch signaling in solid tumors.
Collapse
Affiliation(s)
- Kah Jing Lim
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231, Maryland, USA
| | - William D Brandt
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231, Maryland, USA
| | - Jason A Heth
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, Michigan, USA
| | - Karin M Muraszko
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, Michigan, USA
| | - Xing Fan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, Michigan, USA
| | - Eli E Bar
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231, Maryland, USA.,Department of Neurological Surgery, Case Western University, Cleveland, OH 44106, Ohio, USA
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231, Maryland, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD 21231, Maryland, USA.,Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21231, Maryland, USA
| |
Collapse
|
44
|
Fujihara Y, Ikawa M. GPI-AP release in cellular, developmental, and reproductive biology. J Lipid Res 2015; 57:538-45. [PMID: 26593072 DOI: 10.1194/jlr.r063032] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Indexed: 12/13/2022] Open
Abstract
Glycosylphosphatidylinositol-anchored proteins (GPI-APs) contain a covalently linked GPI anchor located on outer cell membranes. GPI-APs are ubiquitously conserved from protozoa to vertebrates and are critical for physiological events such as development, immunity, and neurogenesis in vertebrates. Both membrane-anchored and soluble GPI-APs play a role in regulating their protein conformation and functional properties. Several pathways mediate the release of GPI-APs from the plasma membrane by vesiculation or cleavage. Phospholipases and putative substrate-specific GPI-AP-releasing enzymes, such as NOTUM, glycerophosphodiesterase 2, and angiotensin-converting enzyme, have been characterized in mammals. Here, the protein modifications resulting from the cleavage of the GPI anchor are discussed in the context of its physiological functions.
Collapse
Affiliation(s)
- Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
45
|
Metrich M, Bezdek Pomey A, Berthonneche C, Sarre A, Nemir M, Pedrazzini T. Jagged1 intracellular domain-mediated inhibition of Notch1 signalling regulates cardiac homeostasis in the postnatal heart. Cardiovasc Res 2015; 108:74-86. [PMID: 26249804 PMCID: PMC4571837 DOI: 10.1093/cvr/cvv209] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 07/23/2015] [Indexed: 12/20/2022] Open
Abstract
Aims Notch1 signalling in the heart is mainly activated via expression of Jagged1 on the surface of cardiomyocytes. Notch controls cardiomyocyte proliferation and differentiation in the developing heart and regulates cardiac remodelling in the stressed adult heart. Besides canonical Notch receptor activation in signal-receiving cells, Notch ligands can also activate Notch receptor-independent responses in signal-sending cells via release of their intracellular domain. We evaluated therefore the importance of Jagged1 (J1) intracellular domain (ICD)-mediated pathways in the postnatal heart. Methods and results In cardiomyocytes, Jagged1 releases J1ICD, which then translocates into the nucleus and down-regulates Notch transcriptional activity. To study the importance of J1ICD in cardiac homeostasis, we generated transgenic mice expressing a tamoxifen-inducible form of J1ICD, specifically in cardiomyocytes. Using this model, we demonstrate that J1ICD-mediated Notch inhibition diminishes proliferation in the neonatal cardiomyocyte population and promotes maturation. In the neonatal heart, a response via Wnt and Akt pathway activation is elicited as an attempt to compensate for the deficit in cardiomyocyte number resulting from J1ICD activation. In the stressed adult heart, J1ICD activation results in a dramatic reduction of the number of Notch signalling cardiomyocytes, blunts the hypertrophic response, and reduces the number of apoptotic cardiomyocytes. Consistently, this occurs concomitantly with a significant down-regulation of the phosphorylation of the Akt effectors ribosomal S6 protein (S6) and eukaryotic initiation factor 4E binding protein1 (4EBP1) controlling protein synthesis. Conclusions Altogether, these data demonstrate the importance of J1ICD in the modulation of physiological and pathological hypertrophy, and reveal the existence of a novel pathway regulating cardiac homeostasis.
Collapse
Affiliation(s)
- Mélanie Metrich
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Rue du Bugnon 27, CH-1011 Lausanne, Switzerland
| | - April Bezdek Pomey
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Rue du Bugnon 27, CH-1011 Lausanne, Switzerland
| | - Corinne Berthonneche
- Cardiovascular Assessment Facility, University of Lausanne, Lausanne, Switzerland
| | - Alexandre Sarre
- Cardiovascular Assessment Facility, University of Lausanne, Lausanne, Switzerland
| | - Mohamed Nemir
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Rue du Bugnon 27, CH-1011 Lausanne, Switzerland
| | - Thierry Pedrazzini
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Rue du Bugnon 27, CH-1011 Lausanne, Switzerland Cardiovascular Assessment Facility, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
46
|
Koga JI, Aikawa M. Application of anti-ligand antibodies to inhibit Notch signaling. Methods Mol Biol 2015; 1187:335-42. [PMID: 25053501 DOI: 10.1007/978-1-4939-1139-4_25] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Emerging evidence suggests that Notch signaling not only regulates biological processes during development but also participates in the pathogenesis of various diseases in adults, including tumor angiogenesis, hematopoietic malignancies, and cardiometabolic syndromes. Notch signaling involves several ligands and receptors that have unique and overlapping functions. Therefore, blocking function of a ligand or receptor with a neutralizing antibody is a useful approach to examine the specific role of each Notch component. In addition, administration of Notch signaling blocking antibodies in experimental animals offers important insights into clinical translation of Notch biology. In this chapter, we describe examples of in vitro and in vivo loss-of-function experiments with blockade of Notch ligands, particularly Delta-like ligand 4 (Dll4).
Collapse
Affiliation(s)
- Jun-ichiro Koga
- The Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB-741J, Boston, MA, 02115, USA
| | | |
Collapse
|
47
|
Doi K, Imai T, Kressler C, Yagita H, Agata Y, Vooijs M, Hamazaki Y, Inoue J, Minato N. Crucial role of the Rap G protein signal in Notch activation and leukemogenicity of T-cell acute lymphoblastic leukemia. Sci Rep 2015; 5:7978. [PMID: 25613394 PMCID: PMC4303867 DOI: 10.1038/srep07978] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 12/23/2014] [Indexed: 01/08/2023] Open
Abstract
The Rap G protein signal regulates Notch activation in early thymic progenitor cells, and deregulated Rap activation (Rap(high)) results in the development of Notch-dependent T-cell acute lymphoblastic leukemia (T-ALL). We demonstrate that the Rap signal is required for the proliferation and leukemogenesis of established Notch-dependent T-ALL cell lines. Attenuation of the Rap signal by the expression of a dominant-negative Rap1A17 or Rap1GAP, Sipa1, in a T-ALL cell line resulted in the reduced Notch processing at site 2 due to impaired maturation of Adam10. Inhibition of the Rap1 prenylation with a geranylgeranyl transferase inhibitor abrogated its membrane-anchoring to Golgi-network and caused reduced proprotein convertase activity required for Adam10 maturation. Exogenous expression of a mature form of Adam10 overcame the Sipa1-induced inhibition of T-ALL cell proliferation. T-ALL cell lines expressed Notch ligands in a Notch-signal dependent manner, which contributed to the cell-autonomous Notch activation. Although the initial thymic blast cells barely expressed Notch ligands during the T-ALL development from Rap(high) hematopoietic progenitors in vivo, the ligands were clearly expressed in the T-ALL cells invading extrathymic vital organs. These results reveal a crucial role of the Rap signal in the Notch-dependent T-ALL development and the progression.
Collapse
Affiliation(s)
- Keiko Doi
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Takahiko Imai
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Christopher Kressler
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Yasutoshi Agata
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Marc Vooijs
- Maastricht Radiation Oncology and School for Oncology and Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| | - Yoko Hamazaki
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Joe Inoue
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Nagahiro Minato
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
48
|
Teodorczyk M, Schmidt MHH. Notching on Cancer's Door: Notch Signaling in Brain Tumors. Front Oncol 2015; 4:341. [PMID: 25601901 PMCID: PMC4283135 DOI: 10.3389/fonc.2014.00341] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/14/2014] [Indexed: 12/20/2022] Open
Abstract
Notch receptors play an essential role in the regulation of central cellular processes during embryonic and postnatal development. The mammalian genome encodes for four Notch paralogs (Notch 1–4), which are activated by three Delta-like (Dll1/3/4) and two Serrate-like (Jagged1/2) ligands. Further, non-canonical Notch ligands such as epidermal growth factor like protein 7 (EGFL7) have been identified and serve mostly as antagonists of Notch signaling. The Notch pathway prevents neuronal differentiation in the central nervous system by driving neural stem cell maintenance and commitment of neural progenitor cells into the glial lineage. Notch is therefore often implicated in the development of brain tumors, as tumor cells share various characteristics with neural stem and progenitor cells. Notch receptors are overexpressed in gliomas and their oncogenicity has been confirmed by gain- and loss-of-function studies in vitro and in vivo. To this end, special attention is paid to the impact of Notch signaling on stem-like brain tumor-propagating cells as these cells contribute to growth, survival, invasion, and recurrence of brain tumors. Based on the outcome of ongoing studies in vivo, Notch-directed therapies such as γ-secretase inhibitors and blocking antibodies have entered and completed various clinical trials. This review summarizes the current knowledge on Notch signaling in brain tumor formation and therapy.
Collapse
Affiliation(s)
- Marcin Teodorczyk
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), Johannes Gutenberg University of Mainz School of Medicine , Mainz , Germany
| | - Mirko H H Schmidt
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), Johannes Gutenberg University of Mainz School of Medicine , Mainz , Germany
| |
Collapse
|
49
|
Stahl R, Schilling S, Soba P, Rupp C, Hartmann T, Wagner K, Merdes G, Eggert S, Kins S. Shedding of APP limits its synaptogenic activity and cell adhesion properties. Front Cell Neurosci 2014; 8:410. [PMID: 25520622 PMCID: PMC4253958 DOI: 10.3389/fncel.2014.00410] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 11/11/2014] [Indexed: 01/05/2023] Open
Abstract
The amyloid precursor protein (APP) plays a central role in Alzheimer's disease (AD) and has essential synapse promoting functions. Synaptogenic activity as well as cell adhesion properties of APP presumably depend on trans-cellular dimerization via its extracellular domain. Since neuronal APP is extensively processed by secretases, it raises the question if APP shedding affects its cell adhesion and synaptogenic properties. We show that inhibition of APP shedding using cleavage deficient forms of APP or a dominant negative α-secretase strongly enhanced its cell adhesion and synaptogenic activity suggesting that synapse promoting function of APP is tightly regulated by α-secretase mediated processing, similar to other trans-cellular synaptic adhesion molecules.
Collapse
Affiliation(s)
- Ronny Stahl
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Department of Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University Munich Munich, Germany
| | - Sandra Schilling
- Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Peter Soba
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Center for Molecular Neurobiology (ZMNH), University of Hamburg Hamburg, Germany
| | - Carsten Rupp
- Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Tobias Hartmann
- Deutsches Institut für DemenzPrävention, Experimental Neurology, Saarland University Homburg/Saar, Germany
| | - Katja Wagner
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Gunter Merdes
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Department of Biosystems Science and Engineering, ETH Zürich Basel, Switzerland
| | - Simone Eggert
- Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Stefan Kins
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany ; Deutsches Institut für DemenzPrävention, Experimental Neurology, Saarland University Homburg/Saar, Germany
| |
Collapse
|
50
|
Pabois A, Devallière J, Quillard T, Coulon F, Gérard N, Laboisse C, Toquet C, Charreau B. The disintegrin and metalloproteinase ADAM10 mediates a canonical Notch-dependent regulation of IL-6 through Dll4 in human endothelial cells. Biochem Pharmacol 2014; 91:510-21. [PMID: 25130545 DOI: 10.1016/j.bcp.2014.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/08/2014] [Accepted: 08/08/2014] [Indexed: 10/24/2022]
Abstract
Although the involvement of the disintegrin and metalloproteinase ADAM10 in several areas of vascular biology is now clearly established, its role in vascular inflammation and in Notch signaling at the endothelial level remains unclear. In this study, we demonstrated that ADAM10 specifically localizes in the CD31(+) endothelial cells (ECs) in normal human cardiac tissues and in cultured primary arterial ECs. In vitro, ADAM10 drives a specific regulation of the Notch pathway in vascular ECs. Using an ADAM10 gain and loss of function approach we show an ADAM10-dependent regulation of Dll1 and Dll4 expression in association with changes in Hes1 and Hey1 expression. We also identified IL-6, IL-8, MCP-1 and sVCAM-1 as novel targets of ADAM10 upon inflammation. Although Notch pathway does not seem to be required for the production of IL-8, MCP-1 and sVCAM-1, the release of IL-6 by ECs occurred through ADAM10 and a canonical Notch signaling pathway, dependent of γ-secretase activity. Moreover, sustained expression of Dll4 mediated by ADAM10 elicits an increased release of IL-6 suggesting a strong implication of the specific Dll4 signaling in this mechanism. Modulation of IL-6 mediated by ADAM10/Notch signaling required PI3K activity. Thus, our findings suggest that ADAM10/Dll4 signaling is a major signaling pathway in ECs driving inflammatory events involved in inflammation and immune cell recruitment.
Collapse
Affiliation(s)
- Angélique Pabois
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM, Université de Nantes, Faculté de Médecine, Nantes F44000, France
| | - Julie Devallière
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM, Université de Nantes, Faculté de Médecine, Nantes F44000, France
| | - Thibaut Quillard
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM, Université de Nantes, Faculté de Médecine, Nantes F44000, France
| | - Flora Coulon
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM, Université de Nantes, Faculté de Médecine, Nantes F44000, France
| | - Nathalie Gérard
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM, Université de Nantes, Faculté de Médecine, Nantes F44000, France
| | - Christian Laboisse
- LUNAM, Université de Nantes, Faculté de Médecine, Nantes F44000, France; Service d'Anatomie Pathologique, CHU de Nantes, Nantes F44000, France
| | - Claire Toquet
- LUNAM, Université de Nantes, Faculté de Médecine, Nantes F44000, France; Service d'Anatomie Pathologique, CHU de Nantes, Nantes F44000, France
| | - Béatrice Charreau
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM, Université de Nantes, Faculté de Médecine, Nantes F44000, France.
| |
Collapse
|